51
|
Abstract
Human studies examining the effects of the dissociative anesthetic ketamine as a model for psychosis and as a rapidly acting antidepressant have spurred great interest in understanding ketamine's actions at molecular, cellular, and network levels. Although ketamine has unequivocal uncompetitive inhibitory effects on N-methyl-d-aspartate receptors (NMDARs) and may preferentially alter the function of NMDARs on interneurons, recent work has questioned whether block of NMDARs is critical for its mood enhancing actions. In this viewpoint, we examine the evolving literature on ketamine supporting NMDARs as important triggers for certain psychiatric effects and the possibility that the antidepressant trigger is unrelated to NMDARs. The rapidly evolving story of ketamine offers great hope for untangling and treating the biology of both depressive and psychotic illnesses.
Collapse
|
52
|
Rame M, Caudal D, Schenker E, Svenningsson P, Spedding M, Jay TM, Godsil BP. Clozapine counteracts a ketamine-induced depression of hippocampal-prefrontal neuroplasticity and alters signaling pathway phosphorylation. PLoS One 2017; 12:e0177036. [PMID: 28472198 PMCID: PMC5417651 DOI: 10.1371/journal.pone.0177036] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 04/20/2017] [Indexed: 01/01/2023] Open
Abstract
Single sub-anesthetic doses of ketamine can exacerbate the symptoms of patients diagnosed with schizophrenia, yet similar ketamine treatments rapidly reduce depressive symptoms in major depression. Acute doses of the atypical antipsychotic drug clozapine have also been shown to counteract ketamine-induced psychotic effects. In the interest of understanding whether these drug effects could be modeled with alterations in neuroplasticity, we examined the impact of acutely-administered ketamine and clozapine on in vivo long-term potentiation (LTP) in the rat’s hippocampus-to-prefrontal cortex (H-PFC) pathway. We found that a low dose of ketamine depressed H-PFC LTP, whereas animals that were co-administrated the two drugs displayed LTP that was similar to a saline-treated control. To address which signaling molecules might mediate such effects, we also examined phosphorylation and total protein levels of GSK3β, GluA1, TrkB, ERK, and mTOR in prefrontal and hippocampal sub-regions. Among the statistically significant effects that were detected (a) both ketamine and clozapine increased the phosphorylation of Ser9-GSK3β throughout the prefrontal cortex and of Ser2481-mTOR in the dorsal hippocampus (DH), (b) clozapine increased the phosphorylation of Ser831-GluA1 throughout the prefrontal cortex and of Ser845-GluA1 in the ventral hippocampus, (c) ketamine treatment increased the phosphorylation of Thr202/Tyr204-ERK in the medial PFC (mPFC), and (d) clozapine treatment was associated with decreases in the phosphorylation of Tyr705-TrkB in the DH and of Try816-TrkB in the mPFC. Further analyses involving phosphorylation effect sizes also suggested Ser831-GluA1 in the PFC displayed the highest degree of clozapine-responsivity relative to ketamine. These results provide evidence for how ketamine and clozapine treatments affect neuroplasticity and signaling pathways in the stress-sensitive H-PFC network. They also demonstrate the potential relevance of H-PFC pathway neuroplasticity for modeling ketamine-clozapine interactions in regards to psychosis.
Collapse
Affiliation(s)
- Marion Rame
- Laboratoire de Physiopathologie des Maladies Psychiatriques, UMR_S894 Inserm, Centre de Psychiatrie et Neurosciences, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Dorian Caudal
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | | | - Per Svenningsson
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Michael Spedding
- Institut de Recherches Servier, Croissy-sur-Seine, France
- Spedding Research Solutions SAS, Le Vesinet, France
| | - Thérèse M. Jay
- Laboratoire de Physiopathologie des Maladies Psychiatriques, UMR_S894 Inserm, Centre de Psychiatrie et Neurosciences, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Bill P. Godsil
- Laboratoire de Physiopathologie des Maladies Psychiatriques, UMR_S894 Inserm, Centre de Psychiatrie et Neurosciences, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- * E-mail:
| |
Collapse
|
53
|
Commemorating John F. MacDonald and the Art of Being a Mentor. Can J Neurol Sci 2016; 43:735-44. [PMID: 27488619 DOI: 10.1017/cjn.2016.272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
John F. MacDonald was a close friend and mentor whose life was ended far too soon on April 22, 2014. To those who knew him, John was an endearing blend of fiery Scotsman, compassionate socialist, dedicated family man, and tireless investigator. Those close to him valued his loyalty and friendship, relished his biting wit, and puzzled at his self-deprecating manner. His career spanned a remarkable period of discovery from the early identification of excitatory amino acid, to the molecular cloning and characterization of glutamate receptors and the elucidation of mechanisms responsible for regulating their function. A true pioneer in each of these areas, John's research has had a lasting impact on our understanding of excitatory synaptic transmission and its plasticity. Our intent in commemorating John's work is to focus on some notable discoveries that highlight the impact and innovative aspects of John's work. In doing so, we also wish to highlight just how greatly our understanding of the glutamate transmitter systems has advanced since the late 1970s, when John first launched his independent neuroscience career.
Collapse
|
54
|
Chauvette S, Soltani S, Seigneur J, Timofeev I. In vivo models of cortical acquired epilepsy. J Neurosci Methods 2016; 260:185-201. [PMID: 26343530 PMCID: PMC4744568 DOI: 10.1016/j.jneumeth.2015.08.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 08/24/2015] [Accepted: 08/26/2015] [Indexed: 10/23/2022]
Abstract
The neocortex is the site of origin of several forms of acquired epilepsy. Here we provide a brief review of experimental models that were recently developed to study neocortical epileptogenesis as well as some major results obtained with these methods. Most of neocortical seizures appear to be nocturnal and it is known that neuronal activities reveal high levels of synchrony during slow-wave sleep. Therefore, we start the review with a description of mechanisms of neuronal synchronization and major forms of synchronized normal and pathological activities. Then, we describe three experimental models of seizures and epileptogenesis: ketamine-xylazine anesthesia as feline seizure triggered factor, cortical undercut as cortical penetrating wound model and neocortical kindling. Besides specific technical details describing these models we also provide major features of pathological brain activities recorded during epileptogenesis and seizures. The most common feature of all models of neocortical epileptogenesis is the increased duration of network silent states that up-regulates neuronal excitability and eventually leads to epilepsy.
Collapse
Affiliation(s)
- Sylvain Chauvette
- Centre de recherche de l'Institut universitaire en santé mentale de Québec (CRIUSMQ), Local F-6500, 2601 de la Canardière, Québec, QC, Canada G1J2G3
| | - Sara Soltani
- Centre de recherche de l'Institut universitaire en santé mentale de Québec (CRIUSMQ), Local F-6500, 2601 de la Canardière, Québec, QC, Canada G1J2G3; Department of Psychiatry and Neuroscience, Université Laval, Québec, Canada
| | - Josée Seigneur
- Centre de recherche de l'Institut universitaire en santé mentale de Québec (CRIUSMQ), Local F-6500, 2601 de la Canardière, Québec, QC, Canada G1J2G3
| | - Igor Timofeev
- Centre de recherche de l'Institut universitaire en santé mentale de Québec (CRIUSMQ), Local F-6500, 2601 de la Canardière, Québec, QC, Canada G1J2G3; Department of Psychiatry and Neuroscience, Université Laval, Québec, Canada.
| |
Collapse
|
55
|
Hoeller AA, Costa APR, Bicca MA, Matheus FC, Lach G, Spiga F, Lightman SL, Walz R, Collingridge GL, Bortolotto ZA, de Lima TCM. The Role of Hippocampal NMDA Receptors in Long-Term Emotional Responses following Muscarinic Receptor Activation. PLoS One 2016; 11:e0147293. [PMID: 26795565 PMCID: PMC4721870 DOI: 10.1371/journal.pone.0147293] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 01/01/2016] [Indexed: 01/28/2023] Open
Abstract
Extensive evidence indicates the influence of the cholinergic system on emotional processing. Previous findings provided new insights into the underlying mechanisms of long-term anxiety, showing that rats injected with a single systemic dose of pilocarpine—a muscarinic receptor (mAChR) agonist—displayed persistent anxiogenic-like responses when evaluated in different behavioral tests and time-points (24 h up to 3 months later). Herein, we investigated whether the pilocarpine-induced long-term anxiogenesis modulates the HPA axis function and the putative involvement of NMDA receptors (NMDARs) following mAChRs activation. Accordingly, adult male Wistar rats presented anxiogenic-like behavior in the elevated plus-maze (EPM) after 24 h or 1 month of pilocarpine injection (150 mg/kg, i.p.). In these animals, mAChR activation disrupted HPA axis function inducing a long-term increase of corticosterone release associated with a reduced expression of hippocampal GRs, as well as consistently decreased NMDAR subunits expression. Furthermore, in another group of rats injected with memantine–an NMDARs antagonist (4 mg/kg, i.p.)–prior to pilocarpine, we found inhibition of anxiogenic-like behaviors in the EPM but no further alterations in the pilocarpine-induced NMDARs downregulation. Our data provide evidence that behavioral anxiogenesis induced by mAChR activation effectively yields short- and long-term alterations in hippocampal NMDARs expression associated with impairment of hippocampal inhibitory regulation of HPA axis activity. This is a novel mechanism associated with anxiety-like responses in rats, which comprise a putative target to future translational studies.
Collapse
Affiliation(s)
- Alexandre A. Hoeller
- Postgraduate Program in Medical Sciences, Center of Health Sciences, University Hospital, Federal University of Santa Catarina, Florianópolis, SC, 88040–970, Brazil
- Department of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, SC, 88040–970, Brazil
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Dorothy Hodgkin Building, Bristol, BS1 3NY, United Kingdom
- * E-mail: (AAH); (TCML)
| | - Ana Paula R. Costa
- Department of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, SC, 88040–970, Brazil
| | - Maíra A. Bicca
- Department of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, SC, 88040–970, Brazil
| | - Filipe C. Matheus
- Department of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, SC, 88040–970, Brazil
| | - Gilliard Lach
- Department of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, SC, 88040–970, Brazil
- Institute of Pharmacology, Innsbruck Medical University, Innsbruck, 6020, Austria
| | - Francesca Spiga
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Bristol, BS1 3NY, United Kingdom
| | - Stafford L. Lightman
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Bristol, BS1 3NY, United Kingdom
| | - Roger Walz
- Postgraduate Program in Medical Sciences, Center of Health Sciences, University Hospital, Federal University of Santa Catarina, Florianópolis, SC, 88040–970, Brazil
- Department of Clinical Medicine, Center of Health Sciences, University Hospital, Federal University of Santa Catarina, Florianópolis, SC, 88040–970, Brazil
| | - Graham L. Collingridge
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Dorothy Hodgkin Building, Bristol, BS1 3NY, United Kingdom
| | - Zuner A. Bortolotto
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Dorothy Hodgkin Building, Bristol, BS1 3NY, United Kingdom
| | - Thereza C. M. de Lima
- Department of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, SC, 88040–970, Brazil
- * E-mail: (AAH); (TCML)
| |
Collapse
|
56
|
McGinnity CJ, Koepp MJ, Hammers A, Riaño Barros DA, Pressler RM, Luthra S, Jones PA, Trigg W, Micallef C, Symms MR, Brooks DJ, Duncan JS. NMDA receptor binding in focal epilepsies. J Neurol Neurosurg Psychiatry 2015; 86:1150-7. [PMID: 25991402 PMCID: PMC4602274 DOI: 10.1136/jnnp-2014-309897] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 04/16/2015] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To demonstrate altered N-methyl-d-aspartate (NMDA) receptor availability in patients with focal epilepsies using positron emission tomography (PET) and [(18)F]GE-179, a ligand that selectively binds to the open NMDA receptor ion channel, which is thought to be overactive in epilepsy. METHODS Eleven patients (median age 33 years, 6 males) with known frequent interictal epileptiform discharges had an [(18)F]GE-179 PET scan, in a cross-sectional study. MRI showed a focal lesion but discordant EEG changes in two, was non-localising with multifocal EEG abnormalities in two, and was normal in the remaining seven patients who all had multifocal EEG changes. Individual patient [(18)F]GE-179 volume-of-distribution (VT) images were compared between individual patients and a group of 10 healthy controls (47 years, 7 males) using Statistical Parametric Mapping. RESULTS Individual analyses revealed a single cluster of focal VT increase in four patients; one with a single and one with multifocal MRI lesions, and two with normal MRIs. Post hoc analysis revealed that, relative to controls, patients not taking antidepressants had globally increased [(18)F]GE-179 VT (+28%; p<0.002), and the three patients taking an antidepressant drug had globally reduced [(18)F]GE-179 VT (-29%; p<0.002). There were no focal abnormalities common to the epilepsy group. CONCLUSIONS In patients with focal epilepsies, we detected primarily global increases of [(18)F]GE-179 VT consistent with increased NMDA channel activation, but reduced availability in those taking antidepressant drugs, consistent with a possible mode of action of this class of drugs. [(18)F]GE-179 PET showed focal accentuations of NMDA binding in 4 out of 11 patients, with difficult to localise and treat focal epilepsy.
Collapse
Affiliation(s)
- C J McGinnity
- Division of Neuroscience, Department of Medicine, Imperial College London, London, UK Medical Research Council Clinical Sciences Centre, London, UK Division of Imaging Sciences & Biomedical Engineering, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - M J Koepp
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK MRI Unit, Epilepsy Society, Chalfont St. Peter, UK
| | - A Hammers
- Division of Neuroscience, Department of Medicine, Imperial College London, London, UK Medical Research Council Clinical Sciences Centre, London, UK Division of Imaging Sciences & Biomedical Engineering, Faculty of Life Sciences & Medicine, King's College London, London, UK Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK MRI Unit, Epilepsy Society, Chalfont St. Peter, UK The Neurodis Foundation, CERMEP Imagerie du Vivant, Lyon, France
| | - D A Riaño Barros
- Division of Neuroscience, Department of Medicine, Imperial College London, London, UK Medical Research Council Clinical Sciences Centre, London, UK
| | - R M Pressler
- Department of Clinical Neurophysiology, Great Ormond Street Hospital for Children NHS Trust, London, UK
| | - S Luthra
- GE Healthcare plc, The Grove Centre, Amersham, UK
| | - P A Jones
- GE Healthcare plc, The Grove Centre, Amersham, UK
| | - W Trigg
- GE Healthcare plc, The Grove Centre, Amersham, UK
| | - C Micallef
- National Hospital for Neurology and Neurosurgery, London, UK
| | - M R Symms
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK MRI Unit, Epilepsy Society, Chalfont St. Peter, UK
| | - D J Brooks
- Division of Neuroscience, Department of Medicine, Imperial College London, London, UK Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - J S Duncan
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK MRI Unit, Epilepsy Society, Chalfont St. Peter, UK
| |
Collapse
|
57
|
Hwa LS, Nathanson AJ, Shimamoto A, Tayeh JK, Wilens AR, Holly EN, Newman EL, DeBold JF, Miczek KA. Aggression and increased glutamate in the mPFC during withdrawal from intermittent alcohol in outbred mice. Psychopharmacology (Berl) 2015; 232:2889-902. [PMID: 25899790 PMCID: PMC4515187 DOI: 10.1007/s00213-015-3925-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 03/20/2015] [Indexed: 12/14/2022]
Abstract
RATIONALE Disrupted social behavior, including occasional aggressive outbursts, is characteristic of withdrawal from long-term alcohol (EtOH) use. Heavy EtOH use and exaggerated responses during withdrawal may be treated using glutamatergic N-methyl-D-aspartate receptor (NMDAR) antagonists. OBJECTIVES The current experiments explore aggression and medial prefrontal cortex (mPFC) glutamate as consequences of withdrawal from intermittent access to EtOH and changes in aggression and mPFC glutamate caused by NMDAR antagonists memantine and ketamine. METHODS Swiss male mice underwent withdrawal following 1-8 weeks of intermittent access to 20 % EtOH. Aggressive and nonaggressive behaviors with a conspecific were measured 6-8 h into EtOH withdrawal after memantine or ketamine (0-30 mg/kg, i.p.) administration. In separate mice, extracellular mPFC glutamate after memantine was measured during withdrawal using in vivo microdialysis. RESULTS At 6-8 h withdrawal from EtOH, mice exhibited more convulsions and aggression and decreased social contact compared to age-matched water controls. Memantine, but not ketamine, increased withdrawal aggression at the 5-mg/kg dose in mice with a history of 8 weeks of EtOH but not 1 or 4 weeks of EtOH or in water drinkers. Tonic mPFC glutamate was higher during withdrawal after 8 weeks of EtOH compared to 1 week of EtOH or 8 weeks of water. Five milligrams per kilogram of memantine increased glutamate in 8-week EtOH mice, but also in 1-week EtOH and water drinkers. CONCLUSIONS These studies reveal aggressive behavior as a novel symptom of EtOH withdrawal in outbred mice and confirm a role of NMDARs during withdrawal aggression and for disrupted social behavior.
Collapse
Affiliation(s)
- Lara S. Hwa
- Tufts University Department of Psychology, Medford, MA 02155
| | | | - Akiko Shimamoto
- Tufts University Department of Psychology, Medford, MA 02155
| | | | | | | | - Emily L. Newman
- Tufts University Department of Psychology, Medford, MA 02155
| | | | - Klaus A. Miczek
- Tufts University Department of Psychology, Medford, MA 02155
- Tufts University Department of Neuroscience, Boston, MA 02111
| |
Collapse
|
58
|
Lodge D, Mercier MS. Ketamine and phencyclidine: the good, the bad and the unexpected. Br J Pharmacol 2015; 172:4254-76. [PMID: 26075331 DOI: 10.1111/bph.13222] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 05/29/2015] [Accepted: 06/03/2015] [Indexed: 12/21/2022] Open
Abstract
The history of ketamine and phencyclidine from their development as potential clinical anaesthetics through drugs of abuse and animal models of schizophrenia to potential rapidly acting antidepressants is reviewed. The discovery in 1983 of the NMDA receptor antagonist property of ketamine and phencyclidine was a key step to understanding their pharmacology, including their psychotomimetic effects in man. This review describes the historical context and the course of that discovery and its expansion into other hallucinatory drugs. The relevance of these findings to modern hypotheses of schizophrenia and the implications for drug discovery are reviewed. The findings of the rapidly acting antidepressant effects of ketamine in man are discussed in relation to other glutamatergic mechanisms.
Collapse
Affiliation(s)
- D Lodge
- Centre for Synaptic Plasticity, School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | - M S Mercier
- Centre for Synaptic Plasticity, School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| |
Collapse
|
59
|
Activation of NMDA receptor by elevated homocysteine in chronic liver disease contributes to encephalopathy. Med Hypotheses 2015; 85:64-7. [DOI: 10.1016/j.mehy.2015.03.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 03/23/2015] [Accepted: 03/28/2015] [Indexed: 11/18/2022]
|
60
|
Honsberger MJ, Taylor JR, Corlett PR. Memories reactivated under ketamine are subsequently stronger: A potential pre-clinical behavioral model of psychosis. Schizophr Res 2015; 164:227-33. [PMID: 25728834 PMCID: PMC4409515 DOI: 10.1016/j.schres.2015.02.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 02/05/2015] [Accepted: 02/08/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Sub-anesthetic doses of the NMDA antagonist ketamine have been shown to model the formation and stability of delusion in human subjects. The latter has been predicted to be due to aberrant prediction error resulting in enhanced destabilization of beliefs. To extend the scope of this model, we investigated the effect of administration of low dose systemic ketamine on memory in a rodent model of memory reconsolidation. METHODS Systemic ketamine was administered either prior to or immediately following auditory fear memory reactivation in rats. Memory strength was assessed by measuring freezing behavior 24h later. Follow up experiments were designed to investigate an effect of pre-reactivation ketamine on short-term memory (STM), closely related memories, and basolateral amygdala (BLA) specific destabilization mechanisms. RESULTS Rats given pre-reactivation, but not post-reactivation, ketamine showed larger freezing responses 24h later compared to vehicle. This enhancement was not observed 3h after the memory reactivation, nor was it seen in a closely related contextual memory. Prior inhibition of a known destabilization mechanism in the BLA blocked the effect of pre-reactivation ketamine. CONCLUSIONS Pre- but not post-reactivation ketamine enhances fear memory. These data together with recent data in human subjects supports a model of delusion fixity that proposes that aberrant prediction errors result in enhanced destabilization and strengthening of delusional belief.
Collapse
Affiliation(s)
- Michael J Honsberger
- Yale University Department of Psychiatry, Division of Molecular Psychiatry, Connecticut Mental Health Center, Abraham Ribicoff Research Facility, 34 Park Street, New Haven 06511, United States
| | - Jane R Taylor
- Yale University Department of Psychiatry, Division of Molecular Psychiatry, Connecticut Mental Health Center, Abraham Ribicoff Research Facility, 34 Park Street, New Haven 06511, United States
| | - Philip R Corlett
- Yale University Department of Psychiatry, Division of Molecular Psychiatry, Connecticut Mental Health Center, Abraham Ribicoff Research Facility, 34 Park Street, New Haven 06511, United States
| |
Collapse
|
61
|
Abstract
NMDA receptors mediate excitatory neurotransmission in brain and spinal cord and play a pivotal role in the neurological disease state of chronic pain, which is caused by central sensitization. Bupivacaine is the indicated local anesthetic in caudal, epidural, and spinal anesthesia and is widely used clinically to manage acute and chronic pain. In addition to blocking Na(+) channels, bupivacaine affects the activity of many other channels, including NMDA receptors. Importantly, bupivacaine inhibits NMDA receptor-mediated synaptic transmission in the dorsal horn of the spinal cord, an area critically involved in central sensitization. We used recombinant NMDA receptors expressed in HEK293 cells and found that increasing concentrations of bupivacaine decreased channel open probability in GluN2 subunit- and pH-independent manner by increasing the mean duration of closures and decreasing the mean duration of openings. Using kinetic modeling of one-channel currents, we attributed the observed current decrease to two main mechanisms: a voltage-dependent "foot-in-the-door" pore block and an allosteric gating effect. Further, the inhibition was state-independent because it occurred to the same degree whether the drug was applied before or after glutamate stimulation and was mediated by extracellular and intracellular inhibitory sites, via hydrophilic and hydrophobic pathways. These results predict that clinical doses of bupivacaine would decrease the peak and accelerate the decay of synaptic NMDA receptor currents during normal synaptic transmission. These quantitative predictions inform possible applications of bupivacaine as preventative and therapeutic approaches in chronic pain.
Collapse
|
62
|
Sivarao DV. The 40-Hz auditory steady-state response: a selective biomarker for cortical NMDA function. Ann N Y Acad Sci 2015; 1344:27-36. [DOI: 10.1111/nyas.12739] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
63
|
Ketamine and other potential glutamate antidepressants. Psychiatry Res 2015; 225:1-13. [PMID: 25467702 DOI: 10.1016/j.psychres.2014.10.028] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Revised: 09/29/2014] [Accepted: 10/28/2014] [Indexed: 11/24/2022]
Abstract
The need for rapid acting antidepressants is widely recognised. There has been much interest in glutamate mechanisms in major depressive disorder (MDD) as a promising target for the development of new antidepressants. A single intravenous infusion of ketamine, a N-methyl-d-aspartate (NMDA) receptor antagonist anaesthetic agent, can alleviate depressive symptoms in patients within hours of administration. The mechanism of action appears to be in part through glutamate release onto non-NMDA receptors including α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and metabotropic receptors. However these are also reported effects on 5-HT, dopamine and intracellular effects on the mammalian target of rapamycin (mTOR) pathway. The effects of SSRI (Selective Serotonin Reuptake Inhibitor) antidepressants may also involve alterations in NMDA function. The article reviews the effect of current antidepressants on NMDA and examines the efficacy and mechanism of ketamine. Response to ketamine is also discussed and comparison with other glutamate drugs including lamotrigine, amantadine, riluzole, memantine, traxoprodil, GLYX-13, MK-0657, RO4917523, AZD2066 and Coluracetam. Future studies need to link the rapid antidepressant effects seen with ketamine to inflammatory theories in MDD.
Collapse
|
64
|
Ruddy RM, Chen Y, Milenkovic M, Ramsey AJ. Differential effects of NMDA receptor antagonism on spine density. Synapse 2014; 69:52-6. [PMID: 25220437 DOI: 10.1002/syn.21784] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 08/27/2014] [Accepted: 08/30/2014] [Indexed: 11/10/2022]
Abstract
The authors demonstrate that different NMDAR antagonists (ketamine and MK-801) have varying effects on spine density depending on dose, drug regimen, and brain region. While acute ketamine treatment increases cortical spine density in mice, subchronic exposure to either drug reduces spine density in both the cortex and striatum.
Collapse
Affiliation(s)
- Rebecca M Ruddy
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada, M5S 1A8
| | | | | | | |
Collapse
|
65
|
Shaffer CL, Osgood SM, Smith DL, Liu J, Trapa PE. Enhancing ketamine translational pharmacology via receptor occupancy normalization. Neuropharmacology 2014; 86:174-80. [PMID: 25063581 DOI: 10.1016/j.neuropharm.2014.07.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 06/10/2014] [Accepted: 07/09/2014] [Indexed: 11/30/2022]
Abstract
Ketamine is used preclinically and clinically to study schizophrenia and depression. Accordingly, it is imperative to understand the temporal relationship between the central concentrations and N-methyl-d-aspartate receptor (NMDAR) interactions of both ketamine and norketamine, its primary active metabolite, across species to assess the translatability of animal models to humans and the back-translation of clinical observations to the preclinical realm. However, such an interspecies normalization of ketamine and norketamine exposures at different clinical and preclinical doses (and their different routes and regimens) is lacking. This work defines the NMDAR occupancy (RO) time course following single doses of ketamine in rats, nonhuman primates (nhp) and humans to allow direct interspecies comparisons of specific ketamine-mediated pharmacodynamics via RO normalization. Total plasma concentration (Cp)-time profiles of ketamine and norketamine were generated from rats and nhp following a single, memory-impairing dose of ketamine; neuropharmacokinetics were determined in rats. [(3)H]MK-801-displacement studies in rats determined estimated mean (95% confidence interval) unbound plasma concentrations (Cp,u) for ketamine and norketamine producing 50% RO (IC50) of 1420 (990, 2140) nM and 9110 (5870, 13700) nM, respectively. Together, these datasets transformed Cp,u-time data to predicted RO (ROpred)-time profiles for rats, nhp and humans at behaviorally relevant ketamine doses. Subsequently, this approach helped determine an infusion paradigm in rats producing a ROpred-time profile mirroring that for a clinically antidepressant infusion. The described indication-independent methodology allows normalization to RO at any time following any ketamine dose (regardless of route or regimen) in any species by simply quantifying the Cp of ketamine and norketamine. Matching temporal RO relationships in animals and humans should allow direct comparisons of specific ketamine-dependent NMDAR-based pharmacodynamics.
Collapse
Affiliation(s)
- Christopher L Shaffer
- Department of Pharmacokinetics, Pharmacodynamics and Metabolism, Worldwide Research & Development, Pfizer Inc., Eastern Point Road, Groton, CT 06340, USA; Neuroscience Research Unit, Worldwide Research & Development, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA.
| | - Sarah M Osgood
- Department of Pharmacokinetics, Pharmacodynamics and Metabolism, Worldwide Research & Development, Pfizer Inc., Eastern Point Road, Groton, CT 06340, USA
| | - Deborah L Smith
- Neuroscience Research Unit, Worldwide Research & Development, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA
| | - JianHua Liu
- Department of Pharmacokinetics, Pharmacodynamics and Metabolism, Worldwide Research & Development, Pfizer Inc., Eastern Point Road, Groton, CT 06340, USA
| | - Patrick E Trapa
- Department of Pharmacokinetics, Pharmacodynamics and Metabolism, Worldwide Research & Development, Pfizer Inc., Eastern Point Road, Groton, CT 06340, USA; Neuroscience Research Unit, Worldwide Research & Development, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA
| |
Collapse
|
66
|
Linsenbardt AJ, Taylor A, Emnett CM, Doherty JJ, Krishnan K, Covey DF, Paul SM, Zorumski CF, Mennerick S. Different oxysterols have opposing actions at N-methyl-D-aspartate receptors. Neuropharmacology 2014; 85:232-42. [PMID: 24878244 DOI: 10.1016/j.neuropharm.2014.05.027] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 05/13/2014] [Accepted: 05/15/2014] [Indexed: 10/25/2022]
Abstract
Oxysterols have emerged as important biomarkers in disease and as signaling molecules. We recently showed that the oxysterol 24(S)-hydroxycholesterol, the major brain cholesterol metabolite, potently and selectively enhances NMDA receptor function at a site distinct from other modulators. Here we further characterize the pharmacological mechanisms of 24(S)-hydroxycholesterol and its synthetic analog SGE201. We describe an oxysterol antagonist of this positive allosteric modulation, 25-hydroxycholesterol. We found that 24(S)-hydroxycholesterol and SGE201 primarily increased the efficacy of NMDAR agonists but did not directly gate the channel or increase functional receptor number. Rather than binding to a direct aqueous-accessible site, oxysterols may partition into the plasma membrane to access the NMDAR, likely explaining slow onset and offset kinetics of modulation. Interestingly, oxysterols were ineffective when applied to the cytosolic face of inside-out membrane patches or through a whole-cell pipette solution, suggesting a non-intracellular site. We also found that another natural oxysterol, 25-hydroxycholesterol, although exhibiting slight potentiation on its own, non-competitively and enantioselectively antagonized the effects of 24(S)-hydroxycholesterol analogs. In summary, we suggest two novel allosteric sites on NMDARs that separately modulate channel gating, but together oppose each other.
Collapse
Affiliation(s)
- Andrew J Linsenbardt
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Amanda Taylor
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christine M Emnett
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Kathiresan Krishnan
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Douglas F Covey
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven M Paul
- Sage Therapeutics, Cambridge, MA 02142, USA; Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10021, USA; Department of Psychiatry, Weill Cornell Medical College, New York, NY 10021, USA; Department of Pharmacology, Weill Cornell Medical College, New York, NY 10021, USA
| | - Charles F Zorumski
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA; Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anatomy & Neurobiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven Mennerick
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA; Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anatomy & Neurobiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
67
|
Binding of ArgTX-636 in the NMDA receptor ion channel. J Mol Biol 2014; 427:176-89. [PMID: 24862283 DOI: 10.1016/j.jmb.2014.05.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 05/12/2014] [Accepted: 05/15/2014] [Indexed: 12/16/2022]
Abstract
The N-methyl-d-aspartate receptors (NMDARs) constitute an important class of ligand-gated cation channels that are involved in the majority of excitatory neurotransmission in the human brain. Compounds that bind in the NMDAR ion channel and act as blockers are use- and voltage-dependent inhibitors of NMDAR activity and have therapeutic potential for treatment of a variety of brain diseases or as pharmacological tools for studies of the neurobiological role of NMDARs. We have performed a kinetic analysis of the blocking mechanism of the prototypical polyamine toxin NMDAR ion channel blocker argiotoxin-636 (ArgTX-636) at recombinant GluN1/2A receptors to provide detailed information on the mechanism of block. The predicted binding site of ArgTX-636 is in the pore region of the NMDAR ion channel formed by residues in the transmembrane M3 and the M2 pore-loop segments of the GluN1 and GluN2A subunits. To assess the predicted binding mode in further detail, we performed an alanine- and glycine-scanning mutational analysis of this pore-loop segment to systematically probe the role of pore-lining M2 residues in GluN1 and GluN2A in the channel block by ArgTX-636. Comparison of M2 positions in GluN1 and GluN2A where mutation influences ArgTX-636 potency suggests differential contribution of the M2-loops of GluN1 and GluN2A to binding of ArgTX-636. The results of the mutational analysis are highly relevant for the future structure-based development of argiotoxin-derived NMDAR channel blockers.
Collapse
|
68
|
Synergistic interaction between ketamine and magnesium in lowering body temperature in rats. Physiol Behav 2014; 127:45-53. [DOI: 10.1016/j.physbeh.2014.01.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 11/20/2013] [Accepted: 01/14/2014] [Indexed: 11/19/2022]
|
69
|
Stuth EAE, Stucke AG, Zuperku EJ. Effects of anesthetics, sedatives, and opioids on ventilatory control. Compr Physiol 2013; 2:2281-367. [PMID: 23720250 DOI: 10.1002/cphy.c100061] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
This article provides a comprehensive, up to date summary of the effects of volatile, gaseous, and intravenous anesthetics and opioid agonists on ventilatory control. Emphasis is placed on data from human studies. Further mechanistic insights are provided by in vivo and in vitro data from other mammalian species. The focus is on the effects of clinically relevant agonist concentrations and studies using pharmacological, that is, supraclinical agonist concentrations are de-emphasized or excluded.
Collapse
Affiliation(s)
- Eckehard A E Stuth
- Medical College of Wisconsin, Anesthesia Research Service, Zablocki VA Medical Center, Milwaukee, Wisconsin, USA.
| | | | | |
Collapse
|
70
|
Wiescholleck V, Manahan-Vaughan D. Long-lasting changes in hippocampal synaptic plasticity and cognition in an animal model of NMDA receptor dysfunction in psychosis. Neuropharmacology 2013; 74:48-58. [DOI: 10.1016/j.neuropharm.2013.01.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 12/21/2012] [Accepted: 01/04/2013] [Indexed: 12/29/2022]
|
71
|
Raffa RB, Pergolizzi JV. Opioid-Induced Hyperalgesia: Is It Clinically Relevant for the Treatment of Pain Patients? Pain Manag Nurs 2013; 14:e67-83. [DOI: 10.1016/j.pmn.2011.04.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Revised: 04/12/2011] [Accepted: 04/13/2011] [Indexed: 11/15/2022]
|
72
|
de la Peña JBI, dela Peña IJI, Lee HL, dela Peña I, Shin CY, Sohn AR, Cheong JH. Pre-exposure to ethanol, but not to caffeine and nicotine, induced place preference and self-administration of the NMDA receptor antagonist-benzodiazepine combination, Zoletil®. Pharmacol Biochem Behav 2013; 110:231-7. [PMID: 23916424 DOI: 10.1016/j.pbb.2013.07.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 07/22/2013] [Accepted: 07/24/2013] [Indexed: 11/30/2022]
Abstract
Zoletil® is an equal amount combination of the NMDA receptor antagonist, tiletamine, and the benzodiazepine, zolazepam, usually used as a veterinary anesthetic. Previous studies have shown that pre-exposure to Zoletil® and other psychoactive drugs (e.g. ketamine, diazepam) plays a significant role in the abuse liability of the compound. However, these studies were only focused on illicit psychoactive drugs and not on the more widely used licit psychoactive substances. Thus, the goal of the present work is to investigate whether pre-exposure to the three most commonly used licit psychoactive substances (caffeine, nicotine, and ethanol) affects the rewarding and reinforcing effects of Zoletil®. Rats were pretreated with caffeine (1.25 or 2.5 mg/kg), nicotine (125 or 250 μg/kg), ethanol (0.5, 2, or 4 g/kg), or saline (1 ml/kg) for 14 days, and evaluated for subsequent Zoletil® place preference (2.5 mg/kg) and self-administration (250 μg/kg). Zoletil® produced neither place preference nor self-administration in saline-pretreated rats. Pre-exposure to caffeine or nicotine does not have significant effects on Zoletil®'s abuse potential. However, pretreatment of ethanol significantly produced Zoletil® place preference and self-administration. These results suggest that individuals who are exposed to ethanol may have a high propensity to use/abuse Zoletil®. More importantly, the present result advocates the careful monitoring on the use and dispensation of Zoletil® or related substances.
Collapse
Affiliation(s)
- June Bryan I de la Peña
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, 26-21 Kongneung-2-dong, Nowon-gu, Seoul 139-742, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
73
|
Jodo E. The role of the hippocampo-prefrontal cortex system in phencyclidine-induced psychosis: a model for schizophrenia. ACTA ACUST UNITED AC 2013; 107:434-40. [PMID: 23792022 DOI: 10.1016/j.jphysparis.2013.06.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 06/05/2013] [Accepted: 06/06/2013] [Indexed: 11/18/2022]
Abstract
Phencyclidine (PCP) is a psychotomimetic drug that induces schizophrenia-like symptoms in healthy individuals and exacerbates pre-existing symptoms in patients with schizophrenia. PCP also induces behavioral and cognitive abnormalities in non-human animals, and PCP-treated animals are considered a reliable pharmacological model of schizophrenia. However, the exact neural mechanisms by which PCP modulates behavior are not known. During the last decade several studies have indicated that disturbed activity of the prefrontal cortex (PFC) may be closely related to PCP-induced psychosis. Systemic administration of PCP produces long-lasting activation of medial PFC (mPFC) neurons in rats, almost in parallel with augmentation of locomotor activity and behavioral stereotypies. Later studies have showed that such PCP-induced behavioral abnormalities are ameliorated by prior administration of drugs that normalize or inhibit excess excitability of PFC neurons. Similar activation of mPFC neurons is not induced by systemic injection of a typical psychostimulant such as methamphetamine, even though behavioral hyperactivity is induced to almost the same level. This suggests that the neural circuits mediating PCP-induced psychosis are different to those mediating methamphetamine-induced psychosis. Locally applied PCP does not induce excitation of mPFC neurons, indicating that PCP-induced tonic excitation of mPFC neurons is mediated by inputs from regions outside the mPFC. This hypothesis is strongly supported by experimental results showing that local perfusion of PCP in the ventral hippocampus, which has dense fiber projections to the mPFC, induces tonic activation of mPFC neurons with accompanying augmentation of behavioral abnormalities. In this review we summarize current knowledge on the neural mechanisms underlying PCP-induced psychosis and highlight a possible involvement of the PFC and the hippocampus in PCP-induced psychosis.
Collapse
Affiliation(s)
- Eiichi Jodo
- Department of Neurophysiology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan.
| |
Collapse
|
74
|
Wild AR, Akyol E, Brothwell SLC, Kimkool P, Skepper JN, Gibb AJ, Jones S. Memantine block depends on agonist presentation at the NMDA receptor in substantia nigra pars compacta dopamine neurones. Neuropharmacology 2013; 73:138-46. [PMID: 23727219 DOI: 10.1016/j.neuropharm.2013.05.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 04/15/2013] [Accepted: 05/08/2013] [Indexed: 11/16/2022]
Abstract
NMDA glutamate receptors (NMDARs) have critical functional roles in the nervous system but NMDAR over-activity can contribute to neuronal damage. The open channel NMDAR blocker, memantine is used to treat certain neurodegenerative diseases, including Parkinson's disease (PD) and is well tolerated clinically. We have investigated memantine block of NMDARs in substantia nigra pars compacta (SNc) dopamine neurones, which show severe pathology in PD. Memantine (10 μM) caused robust inhibition of whole-cell (synaptic and extrasynaptic) NMDARs activated by NMDA at a high concentration or a long duration, low concentration. Less memantine block of NMDAR-EPSCs was seen in response to low frequency synaptic stimulation, while responses to high frequency synaptic stimulation were robustly inhibited by memantine; thus memantine inhibition of NMDAR-EPSCs showed frequency-dependence. By contrast, MK-801 (10 μM) inhibition of NMDAR-EPSCs was not significantly different at low versus high frequencies of synaptic stimulation. Using immunohistochemistry, confocal imaging and stereological analysis, NMDA was found to reduce the density of cells expressing tyrosine hydroxylase, a marker of viable dopamine neurones; memantine prevented the NMDA-evoked decrease. In conclusion, memantine blocked NMDAR populations in different subcellular locations in SNc dopamine neurones but the degree of block depended on the intensity of agonist presentation at the NMDAR. This profile may contribute to the beneficial effects of memantine in PD, as glutamatergic activity is reported to increase, and memantine could preferentially reduce over-activity while leaving some physiological signalling intact.
Collapse
Affiliation(s)
- A R Wild
- Department of Physiology, Development & Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | | | | | | | | | | | | |
Collapse
|
75
|
Sharonova IN, Dvorzhak AY. Blockade of GABAA receptor channels by niflumic acid prevents agonist dissociation. BIOCHEMISTRY (MOSCOW) SUPPLEMENT SERIES A: MEMBRANE AND CELL BIOLOGY 2013. [DOI: 10.1134/s1990747812050169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
76
|
Lester HA, Miwa JM, Srinivasan R. Psychiatric drugs bind to classical targets within early exocytotic pathways: therapeutic effects. Biol Psychiatry 2012; 72:907-15. [PMID: 22771239 PMCID: PMC6167061 DOI: 10.1016/j.biopsych.2012.05.020] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 04/23/2012] [Accepted: 05/21/2012] [Indexed: 11/17/2022]
Abstract
The classical targets for antipsychotic and antidepressant drugs are G protein-coupled receptors and neurotransmitter transporters, respectively. Full therapeutic actions of these drugs require several weeks. We show how therapeutic effects may eventually accrue after existing therapeutic ligands bind to these classical targets, not on the plasma membrane but rather within endoplasmic reticulum (ER) and cis-Golgi. Consequences of such binding may include pharmacological chaperoning: the nascent drug targets are stabilized against degradation and can therefore exit the ER more readily. Another effect may be matchmaking: heterodimers and homodimers of the target form and can more readily exit the ER. Summarizing recent data for nicotinic receptors, we explain how such effects could lead to reduced ER stress and to a decreased unfolded protein response, including changes in gene activation and protein synthesis. In effects not directly related to cellular stress, escorting would allow increased ER exit and trafficking of known associated proteins, as well as other proteins such as growth factors and their receptors, producing both cell-autonomous and non-cell-autonomous effects. Axonal transport of relevant proteins may underlie the several weeks required for full therapy. In contrast, the antidepressant effects of ketamine and other N-methyl-D-aspartate receptor ligands, which occur within <2 hours, could arise from dendritically localized intracellular binding, followed by chaperoning, matchmaking, escorting, and reduced ER stress. Thus, the effects of intracellular binding extend beyond proteostasis of the targets themselves and involve pathways distinct from ion channel and G protein activation. We propose experimental tests and note pathophysiological correlates.
Collapse
Affiliation(s)
- Henry A Lester
- Division of Biology, California Institute of Technology, Pasadena, California.
| | | | | |
Collapse
|
77
|
Graef JD, Benson LC, Sidach SS, Wei H, Lippiello PM, Bencherif M, Fedorov NB. Validation of a high-throughput, automated electrophysiology platform for the screening of nicotinic agonists and antagonists. ACTA ACUST UNITED AC 2012; 18:116-27. [PMID: 22960782 DOI: 10.1177/1087057112457414] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
High-throughput compound screening using electrophysiology-based assays represents an important tool for biomedical research and drug discovery programs. The recent development and availability of devices capable of performing high-throughput electrophysiology-based screening have brought the need to validate these tools by producing data that are consistent with results obtained with conventional electrophysiological methods. In this study, we compared the response properties of hα3β4 and hα4β2 nicotinic receptors to their endogenous ligand acetylcholine (ACh) using three separate electrophysiology platforms: Dynaflow (low-throughput, manual system), PatchXpress 7000A (medium-throughput automated platform), and IonWorks Barracuda (high-throughput automated platform). We found that despite the differences in methodological approaches between these technologies, the EC(50) values from the ACh dose-response curves were consistent between all three platforms. In addition, we have validated the IonWorks Barracuda for both competitive and uncompetitive inhibition assays by using the competitive nicotinic antagonist dihydro-beta-erythroidin (DHβE) and uncompetitive nicotinic antagonist mecamylamine. Furthermore, we have demonstrated the utility of a custom-written algorithm for generating dose-response curves from multiple extrapolated current metrics that allows for discriminating between competitive and uncompetitive inhibition while maintaining high-throughput capacity. This study provides validation of the consistency of results using low-, medium-, and high-throughput electrophysiology platforms and supports their use for screening nicotinic compounds.
Collapse
Affiliation(s)
- John D Graef
- Targacept Inc., Preclinical Department, Winston-Salem, NC, USA.
| | | | | | | | | | | | | |
Collapse
|
78
|
Sani G, Serra G, Kotzalidis GD, Romano S, Tamorri SM, Manfredi G, Caloro M, Telesforo CL, Caltagirone SS, Panaccione I, Simonetti A, Demontis F, Serra G, Girardi P. The role of memantine in the treatment of psychiatric disorders other than the dementias: a review of current preclinical and clinical evidence. CNS Drugs 2012; 26:663-690. [PMID: 22784018 DOI: 10.2165/11634390-000000000-00000] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Memantine, a non-competitive NMDA receptor antagonist approved for Alzheimer's disease with a good safety profile, is increasingly being studied in a variety of non-dementia psychiatric disorders. We aimed to critically review relevant literature on the use of the drug in such disorders. We performed a PubMed search of the effects of memantine in animal models of psychiatric disorders and its effects in human studies of specific psychiatric disorders. The bulk of the data relates to the effects of memantine in major depressive disorder and schizophrenia, although more recent studies have provided data on the use of the drug in bipolar disorder as an add-on. Despite interesting preclinical data, results in major depression are not encouraging. Animal studies investigating the possible usefulness of memantine in schizophrenia are controversial; however, interesting findings were obtained in open studies of schizophrenia, but negative placebo-controlled, double-blind studies cast doubt on their validity. The effects of memantine in anxiety disorders have been poorly investigated, but data indicate that the use of the drug in obsessive-compulsive disorder and post-traumatic stress disorder holds promise, while findings relating to generalized anxiety disorder are rather disappointing. Results in eating disorders, catatonia, impulse control disorders (pathological gambling), substance and alcohol abuse/dependence, and attention-deficit hyperactivity disorder are inconclusive. In most psychiatric non-Alzheimer's disease conditions, the clinical data fail to support the usefulness of memantine as monotherapy or add-on treatment However, recent preclinical and clinical findings suggest that add-on memantine may show antimanic and mood-stabilizing effects in treatment-resistant bipolar disorder.
Collapse
Affiliation(s)
- Gabriele Sani
- NeSMOS Department (Neurosciences, Mental Health, and Sensory Organs), School of Medicine and Psychology, Sapienza University, UOC Psychiatry, SantAndrea Hospital, Rome, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Adolph O, Köster S, Georgieff M, Georgieff EM, Moulig W, Föhr KJ. Promethazine inhibits NMDA-induced currents – New pharmacological aspects of an old drug. Neuropharmacology 2012; 63:280-91. [DOI: 10.1016/j.neuropharm.2012.03.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 02/27/2012] [Accepted: 03/11/2012] [Indexed: 12/24/2022]
|
80
|
Wigestrand MB, Fonnum F, Ivar Walaas S. Subunit-specific modulation of [3H]MK-801 binding to NMDA receptors mediated by dopamine receptor ligands in rodent brain. Neurochem Int 2012; 61:266-76. [DOI: 10.1016/j.neuint.2012.04.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 04/16/2012] [Accepted: 04/18/2012] [Indexed: 11/27/2022]
|
81
|
Cheng YC, Chen KH, Wang JS, Hsu WL, Chien CC, Chen WY, Tsao CW. Rapid analysis of abused drugs using nanostructured silicon surface assisted laser desorption/ionization mass spectrometry. Analyst 2012; 137:654-61. [DOI: 10.1039/c1an15913e] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
82
|
Properties of slow oscillation during slow-wave sleep and anesthesia in cats. J Neurosci 2011; 31:14998-5008. [PMID: 22016533 DOI: 10.1523/jneurosci.2339-11.2011] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Deep anesthesia is commonly used as a model of slow-wave sleep (SWS). Ketamine-xylazine anesthesia reproduces the main features of sleep slow oscillation: slow, large-amplitude waves in field potential, which are generated by the alternation of hyperpolarized and depolarized states of cortical neurons. However, direct quantitative comparison of field potential and membrane potential fluctuations during natural sleep and anesthesia is lacking, so it remains unclear how well the properties of sleep slow oscillation are reproduced by the ketamine-xylazine anesthesia model. Here, we used field potential and intracellular recordings in different cortical areas in the cat to directly compare properties of slow oscillation during natural sleep and ketamine-xylazine anesthesia. During SWS cortical activity showed higher power in the slow/delta (0.1-4 Hz) and spindle (8-14 Hz) frequency range, whereas under anesthesia the power in the gamma band (30-100 Hz) was higher. During anesthesia, slow waves were more rhythmic and more synchronous across the cortex. Intracellular recordings revealed that silent states were longer and the amplitude of membrane potential around transition between active and silent states was bigger under anesthesia. Slow waves were mostly uniform across cortical areas under anesthesia, but in SWS, they were most pronounced in associative and visual areas but smaller and less regular in somatosensory and motor cortices. We conclude that, although the main features of the slow oscillation in sleep and anesthesia appear similar, multiple cellular and network features are differently expressed during natural SWS compared with ketamine-xylazine anesthesia.
Collapse
|
83
|
Lugo-Huitrón R, Blanco-Ayala T, Ugalde-Muñiz P, Carrillo-Mora P, Pedraza-Chaverrí J, Silva-Adaya D, Maldonado PD, Torres I, Pinzón E, Ortiz-Islas E, López T, García E, Pineda B, Torres-Ramos M, Santamaría A, La Cruz VPD. On the antioxidant properties of kynurenic acid: free radical scavenging activity and inhibition of oxidative stress. Neurotoxicol Teratol 2011; 33:538-47. [PMID: 21763768 DOI: 10.1016/j.ntt.2011.07.002] [Citation(s) in RCA: 250] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 06/21/2011] [Accepted: 07/03/2011] [Indexed: 01/27/2023]
Abstract
Kynurenic acid (KYNA) is an endogenous metabolite of the kynurenine pathway for tryptophan degradation and an antagonist of both N-methyl-D-aspartate (NMDA) and alpha-7 nicotinic acetylcholine (α7nACh) receptors. KYNA has also been shown to scavenge hydroxyl radicals (OH) under controlled conditions of free radical production. In this work we evaluated the ability of KYNA to scavenge superoxide anion (O(2)(-)) and peroxynitrite (ONOO(-)). The scavenging ability of KYNA (expressed as IC(50) values) was as follows: OH=O(2)(-)>ONOO(-). In parallel, the antiperoxidative and scavenging capacities of KYNA (0-150 μM) were tested in cerebellum and forebrain homogenates exposed to 5 μM FeSO(4) and 2.5 mM 3-nitropropionic acid (3-NPA). Both FeSO(4) and 3-NPA increased lipid peroxidation (LP) and ROS formation in a significant manner in these preparations, whereas KYNA significantly reduced these markers. Reactive oxygen species (ROS) formation were determined in the presence of FeSO(4) and/or KYNA (0-100 μM), both at intra and extracellular levels. An increase in ROS formation was induced by FeSO(4) in forebrain and cerebellum in a time-dependent manner, and KYNA reduced this effect in a concentration-dependent manner. To further know whether the effect of KYNA on oxidative stress is independent of NMDA and nicotinic receptors, we also tested KYNA (0-100 μM) in a biological preparation free of these receptors - defolliculated Xenopus laevis oocytes - incubated with FeSO(4) for 1 h. A 3-fold increase in LP and a 2-fold increase in ROS formation were seen after exposure to FeSO(4), whereas KYNA attenuated these effects in a concentration-dependent manner. In addition, the in vivo formation of OH evoked by an acute infusion of FeSO(4) (100 μM) in the rat striatum was estimated by microdialysis and challenged by a topic infusion of KYNA (1 μM). FeSO(4) increased the striatal OH production, while KYNA mitigated this effect. Altogether, these data strongly suggest that KYNA, in addition to be a well-known antagonist acting on nicotinic and NMDA receptors, can be considered as a potential endogenous antioxidant.
Collapse
Affiliation(s)
- R Lugo-Huitrón
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, S.S.A., Mexico D.F. 14269, Mexico
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Sensory stimulus evokes inhibition rather than excitation in cerebellar Purkinje cells in vivo in mice. Neurosci Lett 2010; 487:182-6. [PMID: 20965231 DOI: 10.1016/j.neulet.2010.10.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2010] [Revised: 09/28/2010] [Accepted: 10/07/2010] [Indexed: 11/21/2022]
Abstract
Cerebellar Purkinje cells (PC) response precisely to tactile stimulus via granule cells, however, the interaction between sensory evoked synaptic input and the resulting pattern of output spikes in cerebellar cortex is unclear. In this study, we used electrophysiological recording and pharmacological methods to investigate the cerebellar PC in response to natural stimulus on ipsilateral whisker pad in urethane-anesthetized mice. We found that air-puff stimulus on ipsilateral whisker pad evoked neither complex spikes nor simple spike firing, but indeed evoked a strong GABA(A) receptor-mediated inhibition in PCs in cerebellar cortex folium Crus II. Field potential recordings from both molecular layer and PC layer showed that air-puff stimulus evoked a sequence of parallel fiber volley followed by a GABA(A) receptor-mediated inhibition, which completely blocked by AMPA receptor antagonist, NBQX. Cell-attached recordings showed that air-puff stimulus evoked a pause of simple spike firing, GABA(A) receptor antagonist abolished the pause, revealed the tactile stimulus-evoked spike firing in PCs. These results indicated that natural stimulus of whisker pad neither evoked complex spikes, nor fired simple spikes, but induced inhibition in PCs, suggesting that the interneuron network are rapid activated and involved in controlling the spread of sensory information processing in mouse cerebellar cortex folium Crus II.
Collapse
|
85
|
Dinis-Oliveira RJ, Carvalho F, Duarte JA, Dias R, Magalhães T, Santos A. Suicide by hanging under the influence of ketamine and ethanol. Forensic Sci Int 2010; 202:e23-e27. [PMID: 20537829 DOI: 10.1016/j.forsciint.2010.04.047] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Revised: 04/21/2010] [Accepted: 04/28/2010] [Indexed: 11/19/2022]
Abstract
Psychiatric deviations resulting from alcohol and illegal drug abuse may be considered a major risk factor for suicidal behavior. This report describes a suicide by hanging, under the influence of ketamine and alcohol. The victim was a 29-year-old man, found dead hanging by the neck from a metallic beam in the ceiling of his workplace. Besides characteristic signs of hanging seen at the autopsy, toxicological analysis revealed a femoral blood concentration of ketamine and ethanol of 1.3mg/L and 0.66g/L, respectively. Positive qualitative results for ketamine were also detected, in a powder found near the victim and on the victim's nostrils, which suggests nasal inhaling as administration route. The hallucinogenic effects caused by ketamine, associated with an increased sensitivity of N-methyl-d-aspartate (NMDA) receptors to ketamine as result of the previous history of alcoholism should be considered as potential inducing factors in suicide behaviors.
Collapse
|
86
|
Memantine preferentially blocks extrasynaptic over synaptic NMDA receptor currents in hippocampal autapses. J Neurosci 2010; 30:11246-50. [PMID: 20720132 DOI: 10.1523/jneurosci.2488-10.2010] [Citation(s) in RCA: 292] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Glutamate is the major excitatory neurotransmitter in the brain. The NMDA subtype of glutamate receptors (NMDAR) is known to mediate many physiological neural functions. However, excessive activation of NMDARs contributes to neuronal damage in various acute and chronic neurological disorders. To avoid unwanted adverse side effects, blockade of excessive NMDAR activity must therefore be achieved without affecting its physiological function. Memantine, an adamantane derivative, has been used for the treatment of Alzheimer's disease with an excellent clinical safety profile. We previously showed that memantine preferentially blocked neurotoxicity mediated by excessive NMDAR activity while relatively sparing normal neurotransmission, in part because of its uncompetitive antagonism with a fast off-rate. Here, using rat autaptic hippocampal microcultures, we show that memantine at therapeutic concentrations (1-10 microM) preferentially blocks extrasynaptic rather than synaptic currents mediated by NMDARs in the same neuron. We found that memantine blocks extrasynaptic NMDAR-mediated currents induced by bath application of 100 microM NMDA/10 microM glycine with a twofold higher potency than its blockade of the NMDAR component of evoked EPSCs (EPSCs(NMDAR)); this effect persists under conditions of pathological depolarization in the presence of 1 mm extracellular Mg(2+). Thus, our findings provide the first unequivocal evidence to explain the tolerability of memantine based on differential extrasynaptic/synaptic receptor blockade. At therapeutic concentrations, memantine effectively blocks excessive extrasynaptic NMDAR-mediated currents, while relatively sparing normal synaptic activity.
Collapse
|
87
|
Eckle VS, Hucklenbruch C, Todorovic SM. [What do we know about anesthetic mechanisms?: hypnosis, unresponsiveness to surgical incision and amnesia]. Anaesthesist 2010; 58:1144-9. [PMID: 19760252 DOI: 10.1007/s00101-009-1618-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Despite the increase of molecular knowledge in anesthesia research over the past decades there is still ongoing discussion about the mechanisms of anesthesia. This article focuses on presenting anesthetic sensitive ligand and voltage gated ion channels. The impact on anesthetic modulated ion channels is summarized for clinically commonly used anesthetics isoflurane, propofol and ketamine. Furthermore, the anesthetic features hypnosis, unresponsiveness to surgical incision and amnesia and their putative relevant anatomical sites in the central nervous system are briefly introduced.
Collapse
Affiliation(s)
- V-S Eckle
- Department of Anesthesiology, University of Virginia, 22909, Charlottesville, VA, USA.
| | | | | |
Collapse
|
88
|
Control of hippocampal gamma oscillation frequency by tonic inhibition and excitation of interneurons. Nat Neurosci 2009; 13:205-12. [PMID: 20023655 PMCID: PMC2843436 DOI: 10.1038/nn.2464] [Citation(s) in RCA: 166] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Accepted: 11/05/2009] [Indexed: 11/26/2022]
Abstract
Gamma-frequency oscillations depend on phasic synaptic GABAA receptor-mediated inhibition to synchronize spike timing. The spillover of synaptically-released GABA can also activate extrasynaptic GABAA receptors, and such tonic inhibition may also contribute to modulating network dynamics. In many neuronal cell types, tonic inhibition is mediated by δ subunit-containing GABAA receptors. We show that the frequency of in vitro cholinergically-induced gamma oscillations in the mouse hippocampal CA3 region is increased by the activation of NMDA receptors (NMDAR) on interneurons. The NMDAR-dependent increase of gamma oscillation frequency is counteracted by the tonic inhibition of the interneurons mediated by δ subunit-containing GABAA receptors. Recordings of synaptic currents during gamma activity show that NMDAR-mediated increases in oscillation frequency correlate with a progressive synchronization of phasic excitation and inhibition in the network. Thus, the balance between tonic excitation and tonic inhibition of interneurons may modulate gamma frequency by shaping interneuronal synchronization.
Collapse
|
89
|
Portelli J, Aourz N, De Bundel D, Meurs A, Smolders I, Michotte Y, Clinckers R. Intrastrain differences in seizure susceptibility, pharmacological response and basal neurochemistry of Wistar rats. Epilepsy Res 2009; 87:234-46. [PMID: 19833479 DOI: 10.1016/j.eplepsyres.2009.09.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 09/16/2009] [Accepted: 09/18/2009] [Indexed: 10/20/2022]
Abstract
Reliable well-characterised animal models of seizures are necessary in order to better understand the underlying pathophysiological mechanisms as well as to screen potential anticonvulsant drugs. We currently use the focal pilocarpine model as an acute limbic seizure model. Due to breeding problems at the vendor, and apparent changes in pilocarpine-induced seizure susceptibility, we were forced to change breeding locations and vendors over a period of 2 years. Male Wistar rats were either purchased from two breeding locations of Charles River Laboratories (France and Germany), or obtained from Harlan Laboratories (The Netherlands). In the present retrospective study we evaluated the impact of these vendor changes on ketamine dosing to establish anaesthesia, on pilocarpine-induced seizure susceptibility, and on basal extracellular hippocampal noradrenaline, dopamine, serotonin, gamma-amino butyric acid, and glutamate levels of all pilocarpine-treated rats included in our studies. Significant differences were present in all of the parameters analyzed. This study clearly illustrates that intrastrain differences do exist from one vendor/breeding location to another, or even between rats from the same breeding location.
Collapse
Affiliation(s)
- Jeanelle Portelli
- Department of Pharmaceutical Chemistry, Drug Analysis & Drug Information, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | | | | | | | | |
Collapse
|
90
|
Mg2+ imparts NMDA receptor subtype selectivity to the Alzheimer's drug memantine. J Neurosci 2009; 29:2774-9. [PMID: 19261873 DOI: 10.1523/jneurosci.3703-08.2009] [Citation(s) in RCA: 210] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) mediate interneuronal communication and are broadly involved in nervous system physiology and pathology (Dingledine et al., 1999). Memantine, a drug that blocks the ion channel formed by NMDARs, is a widely prescribed treatment of Alzheimer's disease (Schmitt, 2005; Lipton, 2006; Parsons et al., 2007). Research on memantine's mechanism of action has focused on the NMDAR subtypes most highly expressed in adult cerebral cortex, NR1/2A and NR1/2B receptors (Cull-Candy and Leszkiewicz, 2004), and has largely ignored interactions with extracellular Mg(2+) (Mg(2+)(o)). Mg(2+)(o) is an endogenous NMDAR channel blocker that binds near memantine's binding site (Kashiwagi et al., 2002; Chen and Lipton, 2005). We report that a physiological concentration (1 mM) of Mg(2+)(o) decreased memantine inhibition of NR1/2A and NR1/2B receptors nearly 20-fold at a membrane voltage near rest. In contrast, memantine inhibition of the other principal NMDAR subtypes, NR1/2C and NR1/2D receptors, was decreased only approximately 3-fold. As a result, therapeutic memantine concentrations should have negligible effects on NR1/2A or NR1/2B receptor activity but pronounced effects on NR1/2C and NR1/2D receptors. Quantitative modeling showed that the voltage dependence of memantine inhibition also is altered by 1 mM Mg(2+)(o). We report similar results with the NMDAR channel blocker ketamine, a drug used to model schizophrenia (Krystal et al., 2003). These results suggest that currently hypothesized mechanisms of memantine and ketamine action should be reconsidered and that NR1/2C and/or NR1/2D receptors play a more important role in cortical physiology and pathology than previously appreciated.
Collapse
|
91
|
Jung JY, Lee JH, Lee JS, Kim YI. The Effect of a Target Controlled Infusion of Low-Concentration Ketamine on the Heart Rate Variability of Normal Volunteers. Korean J Pain 2009. [DOI: 10.3344/kjp.2009.22.1.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Jai Yun Jung
- Department of Anesthesiology and Pain Medicine, Sanbon Hospital, College of Medicine, Wonkwang University, Gunpo, Korea
| | - Jun Ho Lee
- Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Jeong Seok Lee
- Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Yong Ik Kim
- Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| |
Collapse
|
92
|
Blockade of NMDA receptor channels by 9-aminoacridine and its derivatives. Neurosci Lett 2008; 451:29-33. [PMID: 19111901 DOI: 10.1016/j.neulet.2008.12.036] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2008] [Revised: 12/12/2008] [Accepted: 12/17/2008] [Indexed: 11/21/2022]
Abstract
9-Aminoacridine is known as "foot-in-the-door" NMDA receptor channel blocker because its binding prevents channel closure. Structural determinants of this mechanism of block were studied using a series of 9-aminoacridine derivatives. Experiments were performed on native NMDA receptors of hippocampal pyramidal neurons, isolated from rat brain slices. The use-dependence of block and kinetics of recovery from block were used to characterize mechanism of block produced by the compounds. Modifications, which preserve the flat structure of the tricyclic 9-aminoacridine moiety, affect blocking activity and kinetics but not the foot-in-the-door mechanism. On the contrary, disruption of the flat structure changes the mechanism of block to trapping. It is concluded that flat aromatic structure is one of the critical determinants of the action mechanism of 9-aminoacridine.
Collapse
|
93
|
Gozzi A, Herdon H, Schwarz A, Bertani S, Crestan V, Turrini G, Bifone A. Pharmacological stimulation of NMDA receptors via co-agonist site suppresses fMRI response to phencyclidine in the rat. Psychopharmacology (Berl) 2008; 201:273-84. [PMID: 18704372 DOI: 10.1007/s00213-008-1271-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2008] [Accepted: 07/21/2008] [Indexed: 11/24/2022]
Abstract
RATIONALE Increasing experimental evidence suggests that impaired N-methyl-D: -aspartic acid (NMDA) receptor (NMDAr) function could be a key pathophysiological determinant of schizophrenia. Agonists at the allosteric glycine (Gly) binding site of the NMDA complex can promote NMDAr activity, a strategy that could provide therapeutic efficacy for the disorder. NMDAr antagonists like phencyclidine (PCP) can induce psychotic and dissociative symptoms similar to those observed in schizophrenia and are therefore widely used experimentally to impair NMDA neurotransmission in vivo. OBJECTIVES In the present study, we used pharmacological magnetic resonance imaging (phMRI) to investigate the modulatory effects of endogenous and exogenous agonists at the NMDAr Gly site on the spatiotemporal patterns of brain activation induced by acute PCP challenge in the rat. The drugs investigated were D: -serine, an endogenous agonist of the NMDAr Gly site, and SSR504734, a potent Gly transporter type 1 (GlyT-1) inhibitor that can potentiate NMDAr function by increasing synaptic levels of Gly. RESULTS Acute administration of PCP induced robust and sustained activation of discrete cortico-limbo-thalamic circuits. Pretreatment with D: -serine (1 g/kg) or SSR504734 (10 mg/kg) completely inhibited PCP-induced functional activation. This effect was accompanied by weak but sustained deactivation particularly in cortical areas. CONCLUSIONS These findings suggest that agents that stimulate NMDAr via Gly co-agonist site can potentiate NMDAr activity in the living brain and corroborate the potential for this class of drugs to provide selective enhancement of NMDAr neurotransmission in schizophrenia.
Collapse
Affiliation(s)
- Alessandro Gozzi
- Biology, Neurosciences CEDD, GlaxoSmithKline Medicines Research Centre, Verona, Italy.
| | | | | | | | | | | | | |
Collapse
|
94
|
Stuth EA, Stucke AG, Brandes IF, Zuperku EJ. Anesthetic effects on synaptic transmission and gain control in respiratory control. Respir Physiol Neurobiol 2008; 164:151-9. [DOI: 10.1016/j.resp.2008.05.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2008] [Revised: 05/07/2008] [Accepted: 05/13/2008] [Indexed: 01/09/2023]
|
95
|
|
96
|
N-Methyl-D-Aspartate (NMDA) Antagonists—S(+)-ketamine, Dextrorphan, and Dextromethorphan—Act as Calcium Antagonists on Bovine Cerebral Arteries. J Neurosurg Anesthesiol 2008; 20:241-8. [DOI: 10.1097/ana.0b013e31817f523f] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
97
|
Kalia LV, Kalia SK, Salter MW. NMDA receptors in clinical neurology: excitatory times ahead. Lancet Neurol 2008; 7:742-55. [PMID: 18635022 DOI: 10.1016/s1474-4422(08)70165-0] [Citation(s) in RCA: 309] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Since the N-methyl-D-aspartate receptor (NMDAR) subunits were cloned less than two decades ago, a substantial amount of research has been invested into understanding their physiological function in the healthy CNS. Research has also been directed at their pathological roles in various neurological diseases, including disorders resulting from acute excitotoxic insults (eg, ischaemic stroke, traumatic brain injury), diseases due to chronic neurodegeneration (eg, Alzheimer's, Parkinson's, and Huntington's diseases and amyotrophic lateral sclerosis), disorders arising from sensitisation of neurons (eg, epilepsy, neuropathic pain), and neurodevelopmental disorders associated with NMDAR hypofunction (eg, schizophrenia). Selective NMDAR antagonists have not produced positive results in clinical trials. However, there are other NMDAR-targeted therapies used in current practice that are effective for treating some neurological disorders. In this Review, we describe the evidence for the use of these therapies and provide an overview of drugs being investigated in clinical trials. We also discuss new NMDAR-targeted strategies in clinical neurology.
Collapse
Affiliation(s)
- Lorraine V Kalia
- Division of Neurology, Department of Medicine, University of Toronto, ON, Canada.
| | | | | |
Collapse
|
98
|
Boucetta S, Chauvette S, Bazhenov M, Timofeev I. Focal generation of paroxysmal fast runs during electrographic seizures. Epilepsia 2008; 49:1925-40. [PMID: 18616553 DOI: 10.1111/j.1528-1167.2008.01707.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
PURPOSE A cortically generated Lennox-Gastaut type seizure is associated with spike-wave/poly-spike-wave discharges at 1.0-2.5 Hz and fast runs at 7-16 Hz. Here we studied the patterns of synchronization during runs of paroxysmal fast spikes. METHODS Electrographic activities were recorded using multisite intracellular and field potential recordings in vivo from cats anesthetized with ketamine-xylazine. In different experiments, the recording electrodes were located either at short distances (<1 mm) or at longer distances (up to 12 mm). The main experimental findings were tested in computational models. RESULTS In the majority of cases, the onset and the offset of fast runs occurred almost simultaneously in different recording sites. The amplitude and duration of fast runs could vary by orders of magnitude. Within the fast runs, the patterns of synchronization recorded in different electrodes were as following: (1) synchronous, in phase, (2) synchronous, with phase shift, (3) patchy, repeated in phase/phase shift transitions, and (4) nonsynchronous, slightly different frequencies in different recording sites or absence of oscillatory activity in one of the recording sites; the synchronous patterns (in phase or with phase shifts) were most common. All these patterns could be recorded in the same pair of electrodes during different seizures, and they were reproduced in a computational network model. Intrinsically bursting (IB) neurons fired more spikes per cycle than any other neurons suggesting their leading role in the fast run generation. CONCLUSIONS Once started, the fast runs are generated locally with variable correlations between neighboring cortical foci.
Collapse
Affiliation(s)
- Sofiane Boucetta
- Department of Anatomy and Physiology Laval University, Quebec, Canada
| | | | | | | |
Collapse
|
99
|
Ketamine, but not phencyclidine, selectively modulates cerebellar GABA(A) receptors containing alpha6 and delta subunits. J Neurosci 2008; 28:5383-93. [PMID: 18480294 DOI: 10.1523/jneurosci.5443-07.2008] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Phencyclidine (PCP) and ketamine are dissociative anesthetics capable of inducing analgesia, psychomimetic behavior, and a catatonic state of unconsciousness. Despite broad similarities, there are notable differences between the clinical actions of ketamine and PCP. Ketamine has a lower incidence of adverse effects and generally produces greater CNS depression than PCP. Both noncompetitively inhibit NMDA receptors, yet there is little evidence that these drugs affect GABA(A) receptors, the primary target of most anesthetics. alpha6beta2/3delta receptors are subtypes of the GABA(A) receptor family and are abundantly expressed in granular neurons within the adult cerebellum. Here, using an oocyte expression system, we show that at anesthetically relevant concentrations, ketamine, but not PCP, modulates alpha6beta2delta and alpha6beta3delta receptors. Additionally, at higher concentrations, ketamine directly activates these GABA(A) receptors. Comparatively, dizocilpine (MK-801 [(+)-5-methyl-10,11-dihydro-5H-dibenzo [a,d] cyclohepten-5,10-imine maleate]), a potent noncompetitive antagonist of NMDA receptors that is structurally unrelated to PCP, did not produce any effect on alpha6beta2delta receptors. Of the recombinant GABA(A) receptor subtypes examined (alpha1beta2, alpha1beta2gamma2, alpha1beta2delta, alpha4beta2gamma2, alpha4beta2delta, alpha6beta2gamma2, alpha6beta2delta, and alpha6beta3delta), the actions of ketamine were unique to alpha6beta2delta and alpha6beta3delta receptors. In dissociated granule neurons and cerebellar slice recordings, ketamine potentiated the GABAergic conductance arising from alpha6-containing GABA(A) receptors, whereas PCP showed no effect. Furthermore, ketamine potentiation was absent in cerebellar granule neurons from transgenic functionally null alpha6(-/-) and delta(-/-)mice. These findings suggest that the higher CNS depressant level achieved by ketamine may be the result of its selective actions on alpha6beta2/3delta receptors.
Collapse
|
100
|
The correlation between ketamine and posttraumatic stress disorder in burned service members. ACTA ACUST UNITED AC 2008; 64:S195-8; Discussion S197-8. [PMID: 18376165 DOI: 10.1097/ta.0b013e318160ba1d] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Predisposing factors for posttraumatic stress disorder (PTSD) include experiencing a traumatic event, threat of injury or death, and untreated pain. Ketamine, an anesthetic, is used at low doses as part of a multimodal anesthetic regimen. However, since ketamine is associated with psychosomatic effects, there is a concern that ketamine may increase the risk of developing PTSD. This study investigated the prevalence of PTSD in Operation Iraqi Freedom/Operation Enduring Freedom (OIF/OEF) service members who were treated for burns in a military treatment center. METHODS The PTSD Checklist-Military (PCL-M) is a 17-question screening tool for PTSD used by the military. A score of 44 or higher is a positive screen for PTSD. The charts of all OIF/OEF soldiers with burns who completed the PCL-M screening tool (2002-2007) were reviewed to determine the number of surgeries received, the anesthetic regime used, including amounts given, the total body surface area burned, and injury severity score. Morphine equivalent units were calculated using standard dosage conversion factors. RESULTS The prevalence of PTSD in patients receiving ketamine during their operation(s) was compared with patients not receiving ketamine. Of the 25,000 soldiers injured in OIF/OEF, United States Army Institute of Surgical Research received 603 burned casualties, of which 241 completed the PCL-M. Of those, 147 soldiers underwent at least one operation. Among 119 patients who received ketamine during surgery and 28 who did not; the prevalence of PTSD was 27% (32 of 119) versus 46% (13 of 28), respectively (p = 0.044). CONCLUSIONS Contrary to expectations, patients receiving perioperative ketamine had a lower prevalence of PTSD than soldiers receiving no ketamine during their surgeries despite having larger burns, higher injury severity score, undergoing more operations, and spending more time in the ICU.
Collapse
|